Disease-related stresses trigger hypertrophic growth of the heart that confers markedly increased risk of failure and malignant rhythm disturbance. By contrast, growth of the heart in response to physiological demand is adaptive and not associated with adverse sequalae. Recently, we have shown that FoxO transcription factors play a key role in regulating cardiac myocyte growth. Conversely, the heart is capable of shrinking substantially under a variety of clinically relevant circumstances, such as mechanical support. Muscle atrophy is an active, energy-requiring process requiring activation of ubiquitin ligases. FoxO transcription governs this """"""""atrogene"""""""" response. Thus, FoxO factors are situated at the nexus of multiple forms of cardiac plasticity. Here, we propose a comprehensive analysis of FoxO signaling in cardiac growth and remodeling. Our central hypothesis is that FoxO transcription factors are negative regulators of cardiac growth. We propose to define and manipulate the role of these molecules in 3 major forms of cardiac remodeling: 1) pathological hypertrophy stemming from hemodynamic stress;2) physiological hypertrophy of exercise;and 3) cardiac atrophy from ventricular unloading.
Aim 1 : To test the functional relevance of FoxO transcription factors during cardiac hypertrophy. Using gain-of-function and loss-of function strategies, we will define and manipulate the actions of FoxO in 2 major forms of cardiac growth, viz. pathological and physiological hypertrophy.
Aim 2 : To test the functional relevance of FoxO transcription factors during cardiac atrophy. Using a model of heterotopic cardiac transplantation, we will define and manipulate the actions of FoxO in the setting of ventricular unloading.
Aim 3 : To define and manipulate mechanisms whereby FoxO influences calcineurin and Akt activities, two key mediators of pathological and physiological hypertrophy, respectively. We have evidence that FoxO activation is a robust mechanism of suppressing calcineurin signaling, a major pathway leading to patho- logical cardiac hypertrophy. By contrast, signaling via the PI3K/Akt axis contributes to physiological heart growth, and we have data demonstrating that FoxO is capable of activating Akt. Here, we will decipher mechanisms whereby FoxO targets these major effectors of pathological (calcineurin) and physiological (Akt) cardiac hypertrophy.

Public Health Relevance

Cardiovascular diseases are predicted to be the most common cause of mortality worldwide by the year 2020. Recent studies reveal that FoxO transcription factors are situated at the nexus of multiple forms of cardiac plasticity. By determining their role in pathological cardiac growth and atrophy, we will take steps that may lead to novel strategies to prevent heart failure in humans.

Agency
National Institute of Health (NIH)
Institute
National Heart, Lung, and Blood Institute (NHLBI)
Type
Research Project (R01)
Project #
1R01HL090842-01A1
Application #
7655813
Study Section
Cardiac Contractility, Hypertrophy, and Failure Study Section (CCHF)
Program Officer
Adhikari, Bishow B
Project Start
2009-04-15
Project End
2013-03-31
Budget Start
2009-04-15
Budget End
2010-03-31
Support Year
1
Fiscal Year
2009
Total Cost
$392,500
Indirect Cost
Name
University of Texas Sw Medical Center Dallas
Department
Internal Medicine/Medicine
Type
Schools of Medicine
DUNS #
800771545
City
Dallas
State
TX
Country
United States
Zip Code
75390
Cao, Dian J; Lavandero, Sergio; Hill, Joseph A (2015) Parkin Gone Wild: Unbridled Ubiquitination. Circ Res 117:311-3
Gillette, Thomas G; Hill, Joseph A (2015) Readers, writers, and erasers: chromatin as the whiteboard of heart disease. Circ Res 116:1245-53
Wang, Zhao V; Hill, Joseph A (2015) Protein quality control and metabolism: bidirectional control in the heart. Cell Metab 21:215-226
Daniels, James D; Hill, Joseph A (2015) Funny and late: targeting currents governing heart rate in atrial fibrillation. J Cardiovasc Electrophysiol 26:336-8
Pedrozo, Zully; Criollo, Alfredo; Battiprolu, Pavan K et al. (2015) Polycystin-1 Is a Cardiomyocyte Mechanosensor That Governs L-Type Ca2+ Channel Protein Stability. Circulation 131:2131-42
Luo, Xiang; Hill, Joseph A (2014) Ca²? in the cleft: fast and fluorescent. Circ Res 115:326-8
Bravo-Sagua, Roberto; Torrealba, Natalia; Paredes, Felipe et al. (2014) Organelle communication: signaling crossroads between homeostasis and disease. Int J Biochem Cell Biol 50:55-9
Morales, Cyndi R; Pedrozo, Zully; Lavandero, Sergio et al. (2014) Oxidative stress and autophagy in cardiovascular homeostasis. Antioxid Redox Signal 20:507-18
Parra, Valentina; Verdejo, Hugo E; Iglewski, Myriam et al. (2014) Insulin stimulates mitochondrial fusion and function in cardiomyocytes via the Akt-mTOR-NF?B-Opa-1 signaling pathway. Diabetes 63:75-88
Wang, Zhao V; Li, Dan L; Hill, Joseph A (2014) Heart failure and loss of metabolic control. J Cardiovasc Pharmacol 63:302-13

Showing the most recent 10 out of 62 publications